• Nenhum resultado encontrado

1-Aryl-3-[4-(thieno[3,2-d]pyrimidin-4-yloxy)phenyl]ureas as VEGFR-2 tyrosine kinase inhibitors: Synthesis, biological evaluation, and molecular modelling studies

N/A
N/A
Protected

Academic year: 2021

Share "1-Aryl-3-[4-(thieno[3,2-d]pyrimidin-4-yloxy)phenyl]ureas as VEGFR-2 tyrosine kinase inhibitors: Synthesis, biological evaluation, and molecular modelling studies"

Copied!
10
0
0

Texto

(1)

Volume 2013, Article ID 154856,9pages http://dx.doi.org/10.1155/2013/154856

Research Article

1-Aryl-3-[4-(thieno[3,2-

d]pyrimidin-4-yloxy)phenyl]ureas

as VEGFR-2 Tyrosine Kinase Inhibitors: Synthesis, Biological

Evaluation, and Molecular Modelling Studies

Pedro Soares,

1,2

Raquel Costa,

3

Hugo J. C. Froufe,

4

Ricardo C. Calhelha,

1,4

Daniela Peixoto,

1

Isabel C. F. R. Ferreira,

4

Rui M. V. Abreu,

4

Raquel Soares,

3

and Maria-João R. P. Queiroz

1

1Centro de Qu´ımica, Escola de Ciˆencias, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal 2CIQ/Departamento de Qu´ımica e Bioqu´ımica, Faculdade de Ciˆencias, Universidade do Porto,

Rua do Campo Alegre, 4169-007 Porto, Portugal

3Departamento de Bioqu´ımica (U38-FCT), Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal

4Centro de Investigac¸˜ao de Montanha (CIMO), Escola Superior Agr´aria, Instituto Polit´ecnico de Braganc¸a, Campus de Santa Apol´onia,

Apartado 1172, 5301-855 Braganc¸a, Portugal

Correspondence should be addressed to Maria-Jo˜ao R. P. Queiroz; mjrpq@quimica.uminho.pt Received 7 April 2013; Revised 12 June 2013; Accepted 13 June 2013

Academic Editor: Carmen Domene

Copyright © 2013 Pedro Soares et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. The vascular endothelial growth factor receptor-2 (VEGFR-2) is a tyrosine kinase receptor involved in the growth and differentiation of endothelial cells that are implicated in tumor-associated angiogenesis. In this study, novel 1-aryl-3-[4-(thieno[3,2-d]pyrimidin-4-yloxy)phenyl]ureas were synthesized and evaluated for the VEGFR-2 tyrosine kinase inhibition. Three of these compounds showed good VEGFR-2 inhibition presenting low IC50values (150–199 nM) in enzymatic assays, showing also a significant proliferation inhibition of VEGF-stimulated human umbilical vein endothelial cells (HUVECs) at low concentrations (0.5–1𝜇M), using the Bromodeoxyuridine (BrdU) assay, not affecting cell viability. The determination of the total and phosphorylated (active) VEGFR-2 was performed by western blot, and it was possible to conclude that the compounds significantly inhibit the phosphorylation of the receptor at 1𝜇M pointing to their antiproliferative mechanism of action in HUVECs. The molecular rationale for inhibiting the tyrosine kinase domain of VEGFR-2 was also performed and discussed using molecular docking studies.

1. Introduction

Angiogenesis is the process of new blood vessel formation from preexisting vascular networks by capillary sprouting [1] and plays an important role in the pathogenesis of several disorders including cancer, vasculoproliferative ocular disor-ders, and rheumatoid arthritis [2]. A key regulatory pathway of angiogenesis is mediated by the vascular endothelial growth factor (VEGF), involved in the vascular permeability and an inducer of endothelial cell proliferation, migration, and survival [3], and its cell membrane tyrosine kinase recep-tor VEGFR-2 (also known as KDR) [4]. Upon ligand binding, VEGFR-2 undergoes autophosphorylation, triggering signal-ing pathways leadsignal-ing to endothelial cell proliferation and subsequent angiogenesis. Small molecule inhibitors act by

competing with ATP for its binding site of the VEGFR-2 intracellular tyrosine kinase domain, thereby preventing the signaling pathways that lead to angiogenesis [5]. Several small molecule VEGFR-2 inhibitors have emerged as promising antiangiogenic agents for possible treatment against a wide variety of cancers (Figure 1). Sunitinib was approved for the treatment of renal cell carcinoma and gastrointestinal stromal tumor, and Sorafenib was approved for the treatment of primary kidney cancer and hepatocellular carcinoma [6]. Recently, two new VEGFR-2 inhibitors have been approved for the treatment of advanced renal cell carcinoma: axitinib [7] and pazopanib [8]. A number of thienopyridines and thienopyrimidines ureas have also shown potent VEGFR-2 inhibition activity [9,10] including arylether derivatives [11–

(2)

N H O F HN N H N O N H O N O HN O NH Cl F F F Sunitinib Sorafenib N N N N N NH S O O Pazopanib S N H N N O NH Axitinib CH3 CH3 CH3 CH3 CH3 NH2

Figure 1: Competitive inhibitors of VEGFR-2 in clinical use against several cancers: sunitinib (Sutent, C. P. Pharmaceuticals International, NY, USA), soranefib (Nexavar, Bayer, Germany), axitinib (Inlyta, Pfizer), and pazopanib (Votrient, GlaxoSmithKline).

In this report, we describe an ongoing effort to develop novel small molecules as VEGFR-2 inhibitors, based on the aryletherthieno[3,2-d]pyrimidine arylurea scaffold. The synthesis of the compounds and the VEGFR-2 tyrosine kinase phosphorylation inhibition evaluation using either enzymatic or cellular assays including the determination of the total and of the phosphorylated VEGFR-2 by western blot were performed. The probable binding mode of the 1-aryl-3-[4-(thieno[3,2-d]pyrimidin-4-yloxy)phenyl]ureas with the receptor using docking studies is also presented and dis-cussed.

2. Materials and Methods

2.1. Synthesis. Melting points (∘C) were determined in a

Stuart SMP3 and are uncorrected.1H and13C NMR spectra were recorded on a Varian Unity Plus at 300 and 75.4 MHz, respectively, or on a Bruker Avance III at 400 and 100.6 MHz, respectively. Two-dimensional1H-13C correlations were per-formed to attribute some signals. Mass spectra (MS) EI-TOF or ESI-TOF and HRMS on the M+, [M + H]+ or on [M + Na]+, were performed by the mass spectrometry service of the University of Vigo, C.A.C.T.I., Spain.

The reactions were monitored by thin layer chromatog-raphy (TLC) using Macherey-Nagel precoated aluminium silica gel 60 sheets (0.20 mm) with UV254indicator. Column chromatography was performed on Panreac, silica gel 60, 230–400 mesh.

2.1.1. General Procedure for the Synthesis of Compounds 1a and 1b. In a flask with 5 mL of DMF, thienopyrimidine (1

equiv.), 4-aminophenol (1 equiv.), and K2CO3(4 equiv.) were

heated at 140∘C for 2 h. After cooling, water (5 mL) and ethyl acetate (5 mL) were added. The phases were separated, and the aqueous phase was extracted with more ethyl acetate (2 × 5 mL). The organic phase was dried (MgSO4) and filtered. The solvent was evaporated under reduced pressure giving a solid which was submitted to column chromatography.

4-(Thieno[3,2-d]pyrimidin-4-yloxy)aniline(1a).

4-Chlorothi-eno[3,2-d]pyrimidine (242 mg, 1.42 mmol) and 4-amino-phenol (155 mg, 1.42 mmol) were heated, and the reaction mixture was treated according to the general procedure. Column chromatography using ethyl acetate gave compound

1a as a yellow solid (290 mg, 91%), m.p. 115.8–116.5∘C. 1H NMR (300 MHz, DMSO-𝑑6):𝛿 5.15 (s, 2H, NH2), 6.60 (d,𝐽 = 9.0 Hz, 2H, 2 and 6-H), 6.95 (d,𝐽 = 9.0 Hz, 2H, 3 and 5-H), 7.62 (d,𝐽 = 5.6 Hz, 1H, HetAr), 8.40 (d, 𝐽 = 5.6 Hz, 1H, HetAr), 8.66 (s, 1H, 2󸀠-H) ppm.13C NMR (75.4 MHz, DMSO-𝑑6):𝛿 114.2 (2 and 6-CH), 116.6 (C), 122.3 (3 and 5-CH), 124.1 (CH), 137.1 (CH), 141.7 (C), 146.9 (C), 154.2 (2󸀠-CH), 162.9 (C), 164.3 (C) ppm. MS (EI-TOF)𝑚/𝑧 (%): 243.05 (M+, 100) HRMS (EI-TOF): calcd for C12H9N3OS [M+] 243.0466, found 243.0467.

4-(7-Methylthieno[3,2-d]pyrimidin-4-yloxy)aniline (1b).

4-Chloro-7-methylthieno[3,2-d]pyrimidine (242 mg, 1.31 m mol) and 4-aminophenol (143 mg, 1.31 mmol) were heated, and the reaction mixture was treated according to the general procedure. Column chromatography using ethyl acetate gave compound 1b as a yellow solid (310 mg, 92%), m.p. 167.8–168.6∘C.1H NMR (300 MHz, DMSO-𝑑6):𝛿 2.40 (s, 3H, CH3), 5.13 (s, 2H, NH2), 6.61 (d,𝐽 = 9.0 Hz, 2H, 2 and 6-H), 6.93 (d,𝐽 = 9.0 Hz, 2H, 3 and 5-H), 8.01 (s, 1H, 6󸀠-H), 8.67 (s, 1H, 2󸀠-H) ppm.13C NMR (75.4 MHz, DMSO-𝑑6):𝛿 12.5 (CH3), 114.2 (2 and 6-CH), 116.8 (C), 122.3 (3 and 5-CH),

(3)

131.4 (6󸀠-CH), 132.6 (C), 141.7 (C), 146.8 (C), 153.9 (2󸀠-CH), 161.8 (C), 164.3 (C) ppm. MS (EI-TOF)𝑚/𝑧 (%): 257.06 (M+, 100) HRMS (EI-TOF): calcd for C13H11N3OS [M+] 257.0623, found 257.0621.

2.1.2. General Procedure for the Synthesis of 1,3-Diarylureas

2a–f. Compounds 1a or 1b and different arylisocyanates (1

equiv.) in 6 mL CH2Cl2: THF (1 : 1) were left stirring at room temperature for 16 h. If a precipitate does not come out after this time, hexane (3–5 mL) is added to the mixture to precipitate the product. This was filtered under vacuum to give the corresponding 1,3-diarylureas.

1-Phenyl-3-[4-(thieno[3,2-d]pyrimidin-4-yloxy)phenyl]urea

(2a). From compound 1a (100 mg, 0.410 mmol), and phenylisocyanate (0.0400 mL, 0.410 mmol) compound 2a was isolated as a white solid (117 mg, 79%), m.p. 237.3– 238.7∘C. 1H NMR (400 MHz, DMSO-𝑑6):𝛿 6.95–6.99 (m, 1H, Ar-H), 7.24–7.30 (m, 4H, Ar-H), 7.45–7.47 (m, 2H, 2× Ar-H), 7.54 (d,𝐽 = 9.0 Hz, 2H, 2 × Ar-H), 7.65 (d, 𝐽 = 5.2 Hz, 1H, HetAr), 8.45 (d, 𝐽 = 5.2 Hz, 1H, HetAr), 8.68 (s, 1H, NH), 8.69 (s, 1H,2󸀠󸀠󸀠-H), 8.78 (s, 1H, NH) ppm. 13C NMR (100.6 MHz, DMSO-𝑑6):𝛿 116.8 (C), 118.2 (2 × CH), 119.2 (2× CH), 121.8 (CH), 122.3 (2 × CH), 124.2 (CH), 128.8 (2 × CH), 137.2 (CH), 137.6 (C), 139.6 (C), 146.0 (C), 152.6 (C), 154.1 (2󸀠󸀠󸀠-CH), 163.0 (C), 163.8 (C) ppm. MS (ESI-TOF)𝑚/𝑧

(%): 363.09 ([M + H]+, 100) HRMS (ESI-TOF): calcd for C19H15N4O2S [M + H]+363.0910, found 363.0909.

1-(4-Methoxyphenyl)-3-[4-(thieno[3,2-d]pyrimidin-4-yloxy)

phenyl]urea(2b). From compound 1a (100 mg, 0.410 mmol),

and 4-methoxyphenylisocyanate (0.0500 mL, 0.410 mmol) compound 2b was isolated as a white solid (145 mg, 90%), m.p. 247.2–248.3∘C.1H NMR (400 MHz, DMSO-𝑑6):𝛿 3.71 (s, 3H, OCH3), 6.86 (d, 𝐽 = 9.0 Hz, 2H, 3󸀠 and 5󸀠-H), 7.23 (d, 𝐽 = 9.0 Hz, 2H, 2 × Ar-H), 7.36 (d, 𝐽 = 9.0 Hz, 2H, 2󸀠 and 6󸀠-H), 7.53 (d, 𝐽 = 9.0 Hz, 2H, 2 × Ar-H), 7.65 (d, 𝐽 = 5.6 Hz, 1H, HetAr), 8.44 (d, 𝐽 = 5.6 Hz, 1H, HetAr), 8.50 (s, 1H, NH), 8.69 (s, 1H,2󸀠󸀠󸀠-H), 8.70 (s, 1H, NH) ppm.13C NMR (100.6 MHz, DMSO-𝑑6):𝛿 55.1 (OCH3), 114.0 (3󸀠and 5󸀠-CH), 116.8 (C), 119.1 (2× CH), 120.1 (2󸀠and 6󸀠-CH), 122.3 (2× CH), 124.2 (CH), 132.7 (C), 137.2 (CH), 137.8 (C), 145.9 (C), 152.8 (C), 154.1 (2󸀠󸀠󸀠-CH), 154.5 (C), 163.0 (C), 163.8 (C)

ppm. MS (ESI-TOF)𝑚/𝑧 (%): 393.08 ([M+H]+, 39) HRMS (ESI-TOF): calcd for C20H17N4O3S [M+H]+393.1016, found 393.1026.

1-(4-Cyanophenyl)-3-[4-(thieno[3,2-d]pyrimidin-4-yloxy)

phenyl]urea(2c). From compound 1a (100 mg, 0.410 mmol)

and 4-cyanophenylisocyanate (59.0 mg, 0.410 mmol), com-pound 2c was isolated as a white solid (111 mg, 70%), m.p. 245.5–247.2∘C.1H RMN (300 MHz, DMSO-𝑑6):𝛿 7.26 (d, 𝐽 = 9.2 Hz, 2H, 2× Ar-H), 7.55 (d, 𝐽 = 9.2 Hz, 2H, 2 × Ar-H), 7.62–7.66 (m, 3H, 2󸀠and 6󸀠-H and HetAr), 7.73 (d,𝐽 = 9.2 Hz, 2H, 3󸀠 and 5󸀠-H), 8.45 (d, 𝐽 = 5.2 Hz, 1H, HetAr), 8.69 (s, 1H, 2󸀠󸀠󸀠-H), 8.99 (s, 1H, NH), 9.24 (s, 1H, NH) ppm. 13C NMR (75.4 MHz, DMSO-𝑑6): 𝛿 103.3 (C), 116.8 (C), 118.0 (2󸀠 and 6󸀠-CH), 119.3 (C), 119.7 (2 × CH), 122.4 (2 × CH), 124.3 (CH), 133.3 (3󸀠and 5󸀠-CH), 137.0 (C), 137.2 (CH), 144.2 (C), 146.5 (C), 152.2 (C), 154.1 (2󸀠󸀠󸀠-CH), 163.0 (C), 163.8 (C) ppm. MS (ESI-TOF)𝑚/𝑧 (%): 388.09 ([M + H]+, 100) HRMS (ESI-TOF): calcd for C20H13N5O2S [M + H]+ 388.0863, found 388.0861.

1-Phenyl-3-[4-(7-Methylthieno[3,2-d]pyrimidin-4-yloxy)phe-nyl]urea (2d). From compound 1b (100 mg, 0.389 mmol)

and phenylisocyanate (0.0400 mL, 0.389 mmol), compound

2d was isolated as a white solid (121 mg, 83%), m.p. 285.7–

287.3∘C. 1H NMR (400 MHz, DMSO-𝑑6): 𝛿 2.43 (s, 3H, CH3), 6.96 (m, 1H, Ar-H), 7.23–7.29 (m, 4H, Ar-H), 7.46 (m, 2H, 2× Ar-H), 7.53 (d, 𝐽 = 9.2 Hz, 2H, 2 × Ar-H), 8.10 (s, 1H, 6󸀠󸀠󸀠-H), 8.70 (s, 1H, NH), 8.71 (s, 1H,2󸀠󸀠󸀠-H), 8.78 (s, 1H, NH) ppm.13C NMR (100.6 MHz, DMSO-𝑑6):𝛿 12.5 (CH3), 116.9 (C), 118.2 (2× CH), 119.2 (2 × CH), 121.8 (C), 122.3 (2 × CH), 128.8 (2 × CH), 131.5 (6󸀠󸀠󸀠-CH), 132.7 (C), 137.6 (C), 139.6 (C), 146.1 (C), 152.6 (C), 153.9 (2󸀠󸀠󸀠-CH), 161.9 (C), 163.9 (C) ppm. MS (ESI-TOF)𝑚/𝑧 (%): 377.10 ([M + H]+, 100) HRMS (ESI-TOF): calcd for C20H17N4O2S [M + H]+377.1067, found 377.1064.

1-(4-Methoxyphenyl)-3-[4-(7-methylthieno[3,2-d]pyrimidin-4-yloxy)phenyl]urea (2e). From compound 1b (100 mg,

0.389 mmol) and 4-methoxyphenylisocyanate (0.0500 mL, 0.389 mmol), compound 2e was isolated as a white solid (131 mg, 83%), m.p. 248.4–249.6∘C. 1H NMR (400 MHz, DMSO-𝑑6):𝛿 2.43 (s, 3H, CH3), 3.71 (s, 3H, OCH3), 6.86 (d,𝐽 = 9.2 Hz, 2H, 3󸀠and 5󸀠-H), 7.22 (d,𝐽 = 9.2 Hz, 2H, 3󸀠󸀠 and5󸀠󸀠-H), 7.36 (d,𝐽 = 9.2 Hz, 2H, 2󸀠and 6󸀠-H), 7.52 (d,𝐽 = 9.2 Hz, 2H,2󸀠󸀠 and 6󸀠󸀠-H), 8.06 (s, 1H,6󸀠󸀠󸀠-H), 8.49 (s, 1H, NH), 8.69 (s, 1H, NH), 8.70 (s, 1H,2󸀠󸀠󸀠-H) ppm.13C NMR (100.6 MHz, DMSO-𝑑6): 𝛿 12.5 (CH3), 55.2 (OCH3), 114.0 (3󸀠and 5󸀠-CH), 116.9 (C), 119.1 (2󸀠󸀠and6󸀠󸀠-CH), 120.1 (2󸀠and 6󸀠-CH), 122.2 (3󸀠󸀠 and 5󸀠󸀠-CH), 131.5 (6󸀠󸀠󸀠-CH), 132.7 (C), 137.8 (C), 146.0 (C), 152.8 (C), 153.9 (2󸀠󸀠󸀠-CH), 154.5 (C), 161.9

(C), 163.9 (C) ppm. MS (ESI-TOF) 𝑚/𝑧 (%): 407.12 ([M + H]+, 100) HRMS (ESI-TOF): calcd for C21H19N4O3S [M + H]+407.1172, found 407.1182.

1-(4-Cyanophenyl)-3-[4-(7-methylthieno[3,2-d]pyrimidin-4-yloxy)phenyl]urea (2f ). From compound 1b (100 mg, 0.389

mmol) and 4-cyanophenylisocyanate (56.0 mg, 0.389 mmol), compound 2f was isolated as a white solid (128 mg, 82%), m.p. 246.7–248.2∘C.1H NMR (400 MHz, DMSO-𝑑6):𝛿 2.42 (s, 3H, CH3), 7.25 (d,𝐽 = 9.2 Hz, 2H, 3󸀠󸀠and5󸀠󸀠-H), 7.54 (d, 𝐽 = 9.2 Hz, 2H, 2󸀠󸀠 and 6󸀠󸀠-H), 7.64 (d, 𝐽 = 9.2 Hz, 2H, 2󸀠 and 6󸀠-H), 7.72 (d,𝐽 = 9.2 Hz, 2H, 3󸀠and 5󸀠-H), 8.06 (s, 1H, 6󸀠󸀠󸀠-H), 8.70 (s, 1H,2󸀠󸀠󸀠-H), 8.97 (s, 1H, NH), 9.23 (s, 1H, NH) ppm.13C NMR (100.6 MHz, DMSO-𝑑6):𝛿 12.5 (CH3), 103.3 (C), 117.0 (C), 118.1 (2󸀠and 6󸀠-CH), 119.3 (C), 119.7 (2󸀠󸀠 and 6󸀠󸀠-CH), 122.4 (3󸀠󸀠 and 5󸀠󸀠-CH), 131.5 (6󸀠󸀠󸀠-CH), 132.7 (C), 133.3 (3󸀠 and 5󸀠-CH), 137.0 (C), 144.2 (C), 146.6 (C), 152.2 (C), 153.9 (2󸀠󸀠󸀠-CH), 162.0 (C), 163.8 (C) ppm. MS (ESI-TOF) 𝑚/𝑧 (%): 402.10 ([M + H]+, 100) HRMS (ESI-TOF): calcd for C21H16N5O2S [M + H]+402.1019, found 402.1029.

2.2. VEGFR-2 Enzymatic Inhibition Assay. The compounds

were assessed for VEGFR-2 inhibition activity using the𝑍󸀠 -LYTE-Tyr1 Peptide assay kit (according to the procedures

(4)

N N S O N H N H O N N S O N N S Cl HO N C O 1 2 3 3 2 a d c b 4 1 2 3 6 5 7 2 6 5 3 4 R1 R1= H or CH3 NH2 K2CO3(4 equiv) 2󳰀 4󳰀 6󳰀 7󳰀 R2 NH2 rt, 12 h 1a R1= H, 91% 1b R1= CH3, 92% 2󳰀󳰀󳰀 6󳰀󳰀󳰀 6󳰀󳰀 5󳰀󳰀 3󳰀󳰀 2󳰀󳰀 6󳰀 5󳰀 3󳰀 2󳰀 R1 R2 140∘C, 1 h R1 CH2Cl2: THF (1 : 1), 2a R1= R2= H, 79% 2c R1= H, R2= CN, 70% 2d R1= CH3, R2= H, 83% 2e R1= CH3, R2= OCH3, 83% 2f R1= CH3, R2= CN, 82% DMF, 2b R1= H, R2= OCH3, 90%

Scheme 1: Synthesis of 1-aryl-3-[4-(thieno[3,2-d]pyrimidin-4-yloxy)phenyl]ureas 2a–2f. The yields are presented as % of pure compounds.

recommended by the manufacturer Invitrogen, Cat. PV3190) [14]. Briefly, assays were performed in a total of 20𝜇L in 384-well plates using fluorescence resonance energy transfer technology. A Tyr1 substrate (coumarin-fluorescein double-labeled peptide) at 1.0𝜇M was incubated for 1 h with 4 𝜇g/mL VEGFR-2, 10𝜇M ATP, and inhibitors at room temperature in 50 mM Hepes/Na (pH 7.5), 10 mM MgCl2, 2.0 mM MnCl2, 2.5 mM DTT, 0.10 mM orthovanadate, and 0.01% bovine serum albumin. The wells were incubated at 25∘C for 1 hour and 5.0𝜇L of the development reagent was added to each well. After a second incubation of 1 hour, a stop reagent was added to each well. Using a Biotek FLX800 microplate, the fluorescence was read at 445 nm and 520 nm (excitation 400 nm), and Gen5 Software was used for data analysis. The validation assay was performed using Staurosporine that presented a IC50 value 6 nM that compares to the one reported in the literature (IC507 nM) [14].

2.3. Cell Culture Experiments Using HUVECs

2.3.1. Cell Cultures. Human umbilical vein endothelial cells

(HUVECs) were obtained from the ScienceCell Research Labs (San Diego, CA, USA), HUVECs were seeded on plates coated with 0.2% gelatin (Sigma-Aldrich, Sintra, Por-tugal) and cultured in M199 medium (Sigma-Aldrich, Sin-tra, Portugal) supplemented with 20% fetal bovine serum (FBS) (Invitrogen Life Technologies, Scotland, UK), 1% peni-cillin/streptomycin (Invitrogen Life Technologies), 0.01% heparin (Sigma-Aldrich), and 30 mg/mL endothelial cell growth supplement (Biomedical Technologies Inc., MA, USA) and maintained at 37∘C in a humidified 5% carbon dioxide atmosphere. Cells were kept between passages 3 and 8 for every experiment. The test compounds were dissolved in DMSO and added to cell cultures at a concentration of 0.1–10𝜇M. Incubations were performed for 24 h in medium supplemented with 2% FBS, 1% penicillin/streptomycin, and

60 ng/mL of the vascular endothelial growth factor (Sigma-Aldrich, Sintra, Portugal). Control cells were incubated with vehicle (DMSO at 0.1% in every culture).

2.3.2. MTS Toxicity Assay. HUVECs (2 × 105cells/mL)

were allowed to grow for 24 h and then incubated with the test compounds at a range concentration between 0.1 and 10𝜇M or control (0.1% DMSO) for 24 h. After the incubation period, cells were washed and their viability was assessed using Cell Titer 96 Aqueous ONE Solution Reagent MTS [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium] colorimetric assay (Pro-mega, Madison, USA), according to the instructions provided by the manufacturer and as previously described [15]. Optical density was measured at 492 nm. Three independent experiments were performed, and the results were expressed as mean± SEM.

2.3.3. BrdU Incorporation Assay. HUVECs (6× 104cells/mL)

were grown during 24 h and then were incubated with the compounds at 0.1–10𝜇M or control (0.1% DMSO) for 24 h. Cells were also incubated with 5󸀠-bromodeoxyuridine (BrdU), a thymidine analogue which incorporates into DNA of dividing cells. After incubation with BrdU solution at a final concentration of 0.01 mM during the treatment period, the optical density of proliferating cells (positive for BrdU) after ELISA assay using anti-BrdU-specific antibodies (Roche Diagnostics, Mannheim, Germany) was evaluated at the microplate reader according to the manufacturer’s instruc-tions and as previously reported [16]. The results are given as percentage versus control group (100%).

2.3.4. Cell Apoptosis. HUVECs (6× 104cells/mL) were grown

for 24 h on glass coverslips and incubated with different concentrations (0.1–1𝜇M) of the tested compounds. TUNEL assay was performed using the In Situ Cell Death Detection

(5)

3 2 1 0 C 0.1 0.5 1 5 10 0.1 0.5 1 5 10 0.1 0.5 1 5 10 ∗ ∗ ∗ ∗ ∗ HUVEC via b ili ty Op tical den si ty (492 nm) 2a 2b 2c (a) P ro lif era tin g cells (%) (f o ld incr ea se v er sus co n tr o l) 150 100 50 0 C 0.1 0.5 1 5 10 0.1 0.5 1 5 10 0.1 0.5 1 5 10 2a 2b 2c ∗ ∗ ∗ ∗ ∗ ∗ ∗ ∗ ∗ ∗ ∗ ∗ (b) (f o ld incr ea se v er sus co n tr ol) 150 100 50 0 C 0.1 0.5 1 0.1 0.5 1 0.1 0.5 1 2a 2b 2c ∗ A p o p to tic cells (%) (c)

Figure 2: (a) Cell viability was evaluated in HUVECs upon treatment with 2a, 2b, and 2c at 0.10, 0.50, 1.0, 5.0 and 10 𝜇M, or untreated (control, 0.1% DMSO) using MTS assay. (b) Cell proliferation was assessed by BrdU incorporation assay. Bars represent the percentage of proliferating cells when compared to control group, after incubation with anti-BrdU antibody by ELISA assay. (c) Apoptosis was assessed in HUVECs by TUNEL assay using 2a, 2b, and 2c at 0.1, 0.5, and 1 𝜇M. Bars represent the percentage of apoptotic cells evaluated by the ratio between TUNEL-stained cells and DAPI-stained nuclei in every culture. Results are expressed as mean± SEM of three independent experiments (5 < 𝑛 < 10).∗𝑃 < 0.05 versus control.

kit (Roche Diagnostics, Mannheim, Germany), according to the manufacturer’s instruction and as previously reported [16]. Immunofluorescence was visualized under a fluores-cence microscope (Olympus, BH-2, UK). The percentage of stained cells was evaluated by counting the cells stained with TUNEL (apoptotic cells) divided by the total number of nuclei counterstained with DAPI (Invitrogen, CA, USA) at a 200x magnification field. One thousand nuclei were evaluated. Results were expressed as percentage of control (100%).

2.3.5. Western Blotting Analyses. Proteins were isolated from

HUVEC lysates using RIPA (Chemicon International, CA, USA). Proteins were quantified using a spectrophotometer (Jenway, 6405 UV/vis, Essex, UK), and 20𝜇g of protein were subjected to 8% SDS-PAGE with a 5% stacking gel. After electrophoresis, proteins were blotted into a Hybond nitrocellulose membrane (Amersham, Arlington, VA, USA). Immunodetection for total VEGFR-2 (1 : 1000; Cell Signaling, MA, USA), phosphorylated (activated) VEGFR-2 (1 : 750; Santa Cruz Biotechnology, CA, USA), and𝛽-actin (1 : 3000; Abcam, Cambridge, UK) was accomplished with enhanced chemiluminescence (ECL kit, Amersham, Arlington, USA). The phosphorylated antibody recognizes Tyr951 kinase insert domain, a major site of VEGFR-2 phosphorylation partic-ularly involved in angiogenic processes. The relative inten-sity of each protein blotting analysis was measured using

a computerized software program (Bio-Rad, CA, USA), and the expression of activated and total VEGFR-2 were nor-malized with total VEGFR-2 and𝛽-actin bands, respectively, to compare the expression of proteins in different treatment groups.

2.3.6. Statistical Analyses. All experiments were performed

at least in three independent experiments. Quantifications are expressed as mean ± SEM. Statistical significance of difference between the distinct groups was evaluated by anal-ysis of variance (ANOVA) followed by the Bonferroni test. A difference between experimental groups was considered significant with a confidence interval of 95%, whenever𝑃 < 0.05.

2.4. Docking Simulations Using AutoDock4. A VEGFR-2

crystal structure (PDB: 2XIR) was extracted from the Protein Data Bank (PDB) (http://www.rcsb.org/). The cocrystallized ligand was extracted from the PDB file, and AutoDockTools was used to assign polar hydrogens and Gasteiger charges to the compounds [17]. AutoGrid4 was used to create affinity grid maps for all the atom types. The affinity grids enclosed an area of 100× 100 × 100 with 0.375 ˚A spacing, centered on the coordinates𝑥 = 86.3, 𝑦 = 51.2, and 𝑧 = 48.3. AutoDock4 (version 4.1) with the Lamarckian genetic algorithm was used with the following docking parameters: 100 docking runs, population size of 200, random starting position and

(6)

Table 1: Results of the VEGFR2 enzymatic inhibition assay using compounds 2a–f. Compounds VEGFR-2 IC50(𝜇M)a 2a 0.199 2b 0.188 2c 0.150 2d >100 2e >100 2f >100 aEach IC

50determination is a result of four separate determinations.

conformation, translation step ranges of 2.0 ˚A, mutation rate of 0.02, crossover rate of 0.8, local search rate of 0.06, and 2.5 million energy evaluations [17]. The entire virtual screening experiment was performed on a cluster of 8 Intel Dual-Core 2.8 GHz computers using MOLA software [18]. Inhibition constants(𝐾𝑖) for all ligands were calculated by AutoDock4 as follows:𝐾𝑖= exp((Δ𝐺∗1000)/(𝑅cal∗TK)), where Δ𝐺 is the binding energy,𝑅cal is 1.98719, and TK is 298.15. All figures with structure representations were prepared using PyMOL software [19].

3. Results and Discussion

3.1. Synthesis. We were able to promote the regioselective

attack of the hydroxyl group of the 4-aminophenol in the chlorine nucleophilic displacement of two commercial 4-chlorinated thieno[3,2-d]pyrimidines, using stoichiometric amounts of the reagents, in the synthesis of the aminated compounds 1a and 1b in excellent yields (Scheme 1). This regioselectivity constitutes an important achievement that avoids the reaction of 4-nitrophenols followed by reduction of the nitro compounds to the corresponding amino com-pounds, as often described in the literature [12, 13]. With 4-aminophenol as a reagent, it is possible to use DMF as solvent and heat only at 140∘C instead of diphenylether at 180∘C which is needed for the nucleophilic displacement of the chlorine using 4-nitrophenol. The amino compounds 1a and 1b, obtained in one step, were then reacted with different arylisocyanates to give the corresponding new 1-aryl-3-[4-(thieno[3,2-d]pyrimidin-4-yloxy)phenyl]ureas 2a–2f in high yields (Scheme 1). Compounds 1a, b, and 2a–2f were fully characterized by m.p.,1H,13C NMR, and mass spectrometry including HRMS.

3.2. Biological Evaluation

3.2.1. Enzymatic Assays. The synthesized

thieno[3,2-d]pyr-imidine ureas 2a–f were evaluated for their ability to interact with the VEGFR2 tyrosine kinase domain (Table 1), using an enzymatic FRET-based assay. Compounds 2a, 2b, and 2c displayed the highest inhibition with IC50values of 199, 188, and 150 nM, respectively. The presence of a methoxy group in the para position of the phenyl ring (2b) did not show a significant increase in VEGFR-2 inhibition activity relative to

C 0.1 0.5 1 0.1 0.5 1 0.1 0.5 1 2a 2b 2c (𝜇M) VEGFR-2 𝛽-Actin p VEGFR-2 (a) C 0.1 0.5 1 0.1 0.5 1 0.1 0.5 1 2a 2b 2c Rela ti ve exp re ssio n p VEGFR -2/VEGFR -2 0.6 0.4 0.2 0 ∗ ∗ (b)

Figure 3: (a) Evaluation of phosphorylated and total VEGFR-2 expression in HUVECs after incubation with compounds 2a, 2b, and 2c by western blotting. Representative bands obtained after immunostaining are shown. (b) Quantification of densitometry and mean relative intensity by comparison of the relative intensity of activated VEGFR-2 after normalization with total VEGFR-2 intensity. Data presented as mean ± SEM of two independent experiments.∗𝑃 < 0.05 versus control.

2a, while the presence of a nitrile group in the same position

(2c) promoted a small increase in VEGFR-2 inhibition activity. These findings appear to suggest that the presence of small substituents in the para position of the phenyl group will not have a significant effect on VEGFR-2 potency of the compounds. When a methyl group is present in position 7 of the thieno[3,2-d]pyrimidine moiety (compounds 2d–f), no VEGFR-2 inhibition activity was observed. This finding indicates that substitutions on the mentioned position are not favorable for this series of thieno[3,2-d]pyrimidine ureas, when considering VEGFR-2 inhibition activity.

3.2.2. Cellular Assays. The best compounds in enzymatic

assays (2a, 2b, and 2c) were studied at cellular level. Cell cytotoxicity was first analyzed by MTS assay in the HUVEC cultures upon treatment with 0.10–10𝜇M of compounds 2a–

c (Figure 2(a)). Viability of HUVEC decreased at the highest

concentration of 2c (1.96 ± 0.02 O.D.) and at concentrations of 5.0 and 10𝜇M for 2a (2.00±0.06 O.D. and 1.99±0.03 O.D., resp.) and for 2b (1.84 ± 0.05 O.D. and 1.82 ± 0.04 O.D., resp.) versus control (2.33 ± 0.06 O.D.).

Then, the ability of compounds 2a–c to inhibit VEGF-stimulated proliferation of HUVECs was evaluated using the BrdU incorporation assay (Figure 2(b)). Inhibition of the VEGFR-2 activity was strongly reflected at the cellular level, with all the three compounds showing a statistical significant inhibition activity against VEGF-stimulated HUVEC prolif-eration at 0.5𝜇M (15.2 ± 2.7% for 2a; 12.8 ± 2.4% for 2b,

(7)

LEU840 VAL848 VAL916 CYS919 LEU1035 VAL899 ASP1046 GLU885 LEU889 VAL898 LEU1019 ILE892 ILE888 (a) GLU885 ASP1046 CYS919 (b)

Figure 4: (a) Superimposition of the docking poses at the VEGFR-2 kinase domain for compounds: 2a (cyan), 2b (green) and 2c (orange). (b) Surface representation of the VEGFR-2 kinase hinge with the docking poses of compounds 2c (orange)and 2f (light grey). H-bonds are depicted in dashed yellow lines (distances between 2.9 and 3.3 ˚A) and hydrophobic interactions are depicted in dashed white lines (distances between 3.5 and 4.0 ˚A). Important residues are drawn in sticks. Figures were prepared using PyMOL [19].

and14.7 ± 1.7% for 2c) when comparing to control, which is consider to be100.00 ± 2.8%.

The number of proliferating HUVECs decreased in a dose-dependent manner (Figure 2(b)).

Interestingly, incubation of HUVECs with compounds

2a, 2b, and 2c for 24 h with concentrations 0.1, 0.5, and

1𝜇M (due to cytotoxic effect for higher concentrations,

Figure 2(a)) using the TUNEL assay resulted in an increase

in apoptosis, reaching statistical significance only for 2a at the highest concentration tested (32.8 ± 1.23% increase) comparing with untreated cells (Figure 2(c)).

To investigate the cellular inhibition of VEGFR-2 by the compounds in HUVEC cultures, immunoblotting assays for total and phosphorylated (active) VEGFR-2 were performed

(Figure 3(a)). As illustrated inFigure 3(b), incubation with

compounds 2a and 2b significantly decrease the formation of the active form of the receptor at 1𝜇M concentration, when comparing with the expression of VEGFR-2 (∗𝑃 < 0.05). Compound 2c did not significantly inhibit the activation of the VEGFR-2, although a dose-dependent decrease was observed.

VEGF signaling pathway through the VEGFR-2 acti-vation displays a crucial role in endothelial cells, namely, survival, proliferation, invasion, and apoptosis. Our data sug-gest that compounds 2a–c exert direct actions in HUVECs by inhibiting their growth mainly at 0.5 and 1𝜇M, slightly increasing the percentage of apoptotic cells without cytotoxic effects at the same range of concentrations. The referred cellular-based results are triggered by the inhibition of the phosphorylation (activation) of the VEGFR-2, which is over-expressed in several pathological conditions, such as cancer.

3.3. Molecular Modelling Studies. To better understand the

molecular basis of compounds 2 observed VEGFR-2 inhibi-tion activity, docking simulainhibi-tions using AutoDock4 [17] were

carried out against the VEGFR-2 kinase domain (PDB code 2XIR).

Figure 4(a) displays the superimposition of the best

docked poses obtained for the most potent

thieno[3,2-d]pyrimidine derivatives 2a–c. They present similar docking

poses, with the binding conformation stabilized mainly by the formation of a network of four H-bonds. The two N– H of the urea group form two H-bonds with the Glu885 carboxyl group, while the C=O bond of the urea group forms a third H-bond with the backbone N–H of Asp1046. These H-bonds are similar to the ones observed with other known VEGFR-2 inhibitors presenting a biphenyl urea moiety, including the drug Soranefib (Figure 1). A fourth H-bond is predicted between the N atom in position 4 of the

thieno[3,2-d]pyrimidine moiety and the backbone N–H of Cys919.

The terminal aryl group is positioned in a hydropho-bic region formed by several hydrophohydropho-bic residues (Ile888, Leu889, Ile892, Val898, Val 899, and Leu1019). From

Figure 4(a), it is clearly observed that the 4-methoxyphenyl

and 4-cyanophenyl substituents occupy part of a large hydrophobic pocket that is able to accommodate them, providing an explanation for compounds 2b and 2c being almost equipotent to 2a that bears no substituents in the phenyl ring. Substitutions of hydrophobic groups in ortho or meta positions of the phenyl ring will probably be worth exploring for synthesis of even more potent compounds in this series.

The lack of inhibition by compounds 2d–f for VEGFR-2 was analysed by superimposing the docked pose of compound 2c (the most potent in the enzymatic assays, 150 nM) and the corresponding 7-methylated compound

2f (Figure 4(b)). The methyl group seems to displace the

thieno[3,2-d]pyrimidine ring away from Cys919. This displacement probably interferes with the formation of the H-bond between the N atom in position 4 of the

(8)

thieno[3,2-d]pyrimidine ring and the backbone N–H of Cys919 (Figure 4(b)). These results demonstrate that this H-bond is an absolute requirement for VEGFR-2 inhibition potency of this thieno[3,2-d]pyrimidine series, and the methyl group in position 7 of compounds 2d–f promotes loss of VEGFR-2 inhibition activity by impairing H-bond formation. We can also infer that other substitutions in position 7 of the thieno[3,2-b]pyridine are probably not desirable as they will also impair H-bond formation.

4. Conclusion

In conclusion, we reported the synthesis of novel 1-aryl-3-[4-(thieno[3,2-d]pyrimidin-4-yloxy)phenyl]ureas 2a–2f

in high yields in two steps: the regioselective nucleophilic displacement of chlorine on two

4-chlorothieno[3,2-d]pyrimidines using 4-aminophenol and the reaction of the

resultant 4-(thieno[3,2-d]pyrimidin-4-yloxy)anilines with arylisocyanates. Compounds 2a–c, without the methyl group in position 7, were shown to be the most potent VEGFR-2 inhibitors presenting IC50 values between 150–199 nM in enzymatic assays. The cell culture assays enable us to identify the angiogenic steps targeted by the compounds 2a–c. In fact, compounds were evaluated using a cell-based HUVEC assay and showed activity at the same concentration range (0.5–1𝜇M) without affecting cell viability, inhibiting cell proliferation, and avoiding the formation of active VEGFR-2 specially in the case of compounds 2a and 2b, thus showing their anti-angiogenic potential. Compound 2a increases also apoptosis at the highest concentration tested (1𝜇M) using the TUNEL assay. By performing docking studies, the binding conformation and H-bond network of the most promising compounds 2a–c were predicted. Based on these molecular modelling studies, several suggestions can be done for the synthesis of even more potent compounds.

Conflict of Interests

The authors declare that they have no conflict of interests.

Acknowledgments

The authors would like to acknowledge Foundation for the Science and Technology (FCT Portugal) for financial support through the NMR Portuguese network (Bruker 400 Avance III-Univ Minho). FCT and European Fund for Regional Development (FEDER)-COMPETE-QREN-EU for financial support through the research unities C/QUI/UI686/2011, OE/SAU/UI0038/2011, and PEst-OE/AGR/UI0690/2011, the research project PTDC/QUI-QUI/111060/2009, and the post-Doctoral Grant attributed to Ricardo C. Calhelha (SFRH/BPD/68344/2010) also financed by Human Potential Operational Programme (POPH) and Social European Fund (FSE).

References

[1] S. Patan, “Vasculogenesis and angiogenesis as mechanisms of vascular network formation, growth and remodeling,” Journal

of Neuro-Oncology, vol. 50, no. 1-2, pp. 1–15, 2000.

[2] J. Folkman and Y. Shing, “Angiogenesis,” The Journal of

Biologi-cal Chemistry, vol. 267, no. 16, pp. 10931–10934, 1992.

[3] N. Ferrara, H.-P. Gerber, and J. LeCouter, “The biology of VEGF and its receptors,” Nature Medicine, vol. 9, no. 6, pp. 669–676, 2003.

[4] M. A. Lemmon and J. Schlessinger, “Cell signaling by receptor tyrosine kinases,” Cell, vol. 141, no. 7, pp. 1117–1134, 2010. [5] S. Baka, A. R. Clamp, and G. C. Jayson, “A review of the

latest clinical compounds to inhibit VEGF in pathological angiogenesis,” Expert Opinion on Therapeutic Targets, vol. 10, no. 6, pp. 867–876, 2006.

[6] A. Eichholz, S. Merchant, and A. M. Gaya, “Anti-angiogenesis therapies: their potential in cancer management,” Oncology

Targets Therapy, vol. 3, pp. 69–82, 2010.

[7] D. D. Hu-Lowe, H. Y. Zou, M. L. Grazzini et al., “Nonclinical antiangiogenesis and antitumor activities of axitinib (AG-013736), an oral, potent, and selective inhibitor of vascular endothelial growth factor receptor tyrosine kinases 1, 2, 3,”

Clinical Cancer Research, vol. 14, no. 22, pp. 7272–7283, 2008.

[8] L. Cowey, G. Sonpavde, and T. E. Hutson, “New advancements and developments in treatment of renal cell carcinoma: focus on pazopanib,” OncoTargets and Therapy, vol. 3, pp. 147–155, 2010. [9] Y. Dai, Y. Guo, R. R. Frey et al., “Thienopyrimidine ureas

as novel and potent multitargeted receptor tyrosine kinase inhibitors,” Journal of Medicinal Chemistry, vol. 48, no. 19, pp. 6066–6083, 2005.

[10] H. R. Heyman, R. R. Frey, P. F. Bousquet et al., “Thienopyridine urea inhibitors of KDR kinase,” Bioorganic and Medicinal

Chemistry Letters, vol. 17, no. 5, pp. 1246–1249, 2007.

[11] S. Claridge, F. Raeppel, M.-C. Granger et al., “Discovery of a novel and potent series of thieno[3,2-b]pyridine-based inhibitors of c-Met and VEGFR2 tyrosine kinases,” Bioorganic

and Medicinal Chemistry Letters, vol. 18, no. 9, pp. 2793–2798,

2008.

[12] S. Raeppel, S. Claridge, O. Saavedra et al., “N-(3-fluoro- 4-(2-arylthieno[3,2-b]pyridin-7-yloxy)phenyl)-2-oxo-3-phenylimidazolidine-1-carboxamides: a novel series of dual c-Met/VEGFR2 receptor tyrosine kinase inhibitors,” Bioorganic

and Medicinal Chemistry Letters, vol. 19, no. 5, pp. 1323–1328,

2009.

[13] O. Saavedra, S. Claridge, L. Zhan et al., “N3-Arylmalonamides: a new series of thieno[3,2-b]pyridine based inhibitors of c-Met and VEGFR2 tyrosine kinases,” Bioorganic and Medicinal

Chemistry Letters, vol. 19, no. 24, pp. 6836–6839, 2009.

[14] “Staurosporine IC50 calculated by Invitrogen with the same FRET-based VEGFR-2 enzymatic kit used in this study,” http://tools.invitrogen.com.

[15] R. Costa, ˆA. Carneiro, A. Rocha et al., “Bevacizumab and ranibizumab on microvascular endothelial cells: a comparative study,” Journal of Cellular Biochemistry, vol. 108, no. 6, pp. 1410– 1417, 2009.

[16] A. Rocha, I. Azevedo, and R. Soares, “Anti-angiogenic effects of imatinib target smooth muscle cells but not endothelial cells,”

Angiogenesis, vol. 10, no. 4, pp. 279–286, 2007.

[17] G. M. Morris, H. Ruth, W. Lindstrom et al., “Software news and updates AutoDock4 and AutoDockTools4: automated docking with selective receptor flexibility,” Journal of Computational

Chemistry, vol. 30, no. 16, pp. 2785–2791, 2009.

[18] R. M. V. Abreu, H. J. C. Froufe, M. J. R. P. Queiroz, and I. C. F. R. Ferreira, “MOLA: a bootable, self-configuring system for virtual screening using AutoDock4/Vina on computer clusters,”

(9)

[19] “The PyMOL Molecular Graphics System, Version 1.3, Schr¨odinger, LLC,” September 2012,http://www.pymol.org/.

(10)

Submit your manuscripts at

http://www.hindawi.com

Enzyme Research

Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013

Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013

Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013

Advances in

Virology

ISRN

Biotechnology Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013

Archaea

Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013

Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013

Bioinformatics

Advances in

Hindawi Publishing Corporation http://www.hindawi.com

Genomics

International Journal of Volume 2013 ISRN Microbiology Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013 Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013

Signal Transduction

Journal of Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013

Peptides

International Journal of

ISRN

Zoology Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013

Stem Cells International

Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013

ISRN

Cell Biology Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013

BioMed Research International

Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013

Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013 Biochemistry Research International

Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013 Journal of

Marine Biology

Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013

Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013

The Scientific

World Journal

ISRN

Molecular Biology Hindawi Publishing Corporation

http://www.hindawi.com Volume 2013

International Journal of

Evolutionary Biology

Hindawi Publishing Corporation

Referências

Documentos relacionados

Parágrafo único – Os empregados não contemplados nos pisos mínimos salariais descritos na cláusula terceira ficam assegurados a estes o reajuste negociado na cláusula quarta,

CUIDADO: Se for colocado um filtro a jusante da bomba de êmbolo, e em função do filtro de cola utilizado (com válvula de esfera), a pressão da cola não será completamente

Uma  vez  que  a  correção  de  fenótipos  diminui  a  contribuição do parentesco para a análise, pressupõe- se que a metodologia RR-Blup foi a mais penalizada,

UNIBANCO ARTEPLEX RICARDO GRUPO ESPAÇO / ADHEMAR SÃO PAULO SP 2 1 SP CURITIBA - ARTEPLEX COTLOG GRUPO ESPAÇO / ADHEMAR CURITIBA PR SP DRAGÃO DO MAR COTLOG GRUPO ESPAÇO /

O presente trabalho aplicará a simulação computacional em Simulink através do modelamento dinâmico de um dos componentes do chiller de absorção de simples efeito - o

Outro ponto crítico para o fechamento controlado de disjuntores de transformadores de potência sem carga é que o fluxo prospectivo criado pela tensão no instante do fechamento

A adaptação com a sociedade paulistana foi um processo que costumou levar pouco tempo segundo os entrevistados, tanto que foi observada a intenção de seguir mantendo

d. Conta 63: Gastos com o pessoal. A Requerente, para justificar a sua tese, refere que, após a análise do extrato da conta 231, verifica-se que foram efetuados os